Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Obes Res Clin Pract ; 14(2): 151-157, 2020.
Article in English | MEDLINE | ID: mdl-32061582

ABSTRACT

BACKGROUND: The association between anthropometric variables and cardiovascular disease (CVD) risk among Africans is unclear. We examined the discriminative ability of anthropometric variables and estimate cutoffs for predicting CVD risk among Africans. METHODS: The Research on Obesity and Diabetes among African Migrants (RODAM) study was a multisite cross-sectional study of Africans in Ghana and Europe. We calculated AHA/ACC Pooled Cohort Equations (PCE) scores for 3661 participants to ascertain CVD risk, and compared a body shape index (ABSI), body mass index (BMI), waist circumference (WC), waist-to-hip ratio (WHR), Relative Fat Mass (RFM), and Waist to Height Ratio (WHtR). Logistic regression and receiver operating curve analyses were performed to derive cutoffs for identifying high predicted CVD risk (PCE score ≥7.5%). RESULTS: Among men, WC (adjusted Odds Ratio (aOR): 2.25, 95% CI; 1:50-3:37) was strongly associated with CVD risk. Among women, WC (aOR: 1.69, 95% CI: 1:33-2:14) also displayed the strongest association with CVD risk in the BMI-adjusted model but WHR displayed the strongest fit. All variables were superior discriminators of high CVD risk in men (c-statistic range: 0.887-0.891) than women (c-statistic range: 0.677-0.707). The optimal WC cutoff for identifying participants at high CVD risk was 89 cm among men and identified the most cases (64%). Among women, the recommended WC cutoff of 94 cm or WHR cutoff of 0.90 identified the most cases (92%). CONCLUSIONS: Anthropometric variables were stronger discriminators of high CVD risk in African men than women. Greater WC was associated with high CVD risk in men while WHR and WC were associated with high CVD risk in women.


Subject(s)
Anthropometry , Black People/statistics & numerical data , Cardiovascular Diseases/ethnology , Obesity/ethnology , Risk Assessment/ethnology , Adipose Tissue , Body Mass Index , Cardiovascular Diseases/etiology , Cross-Sectional Studies , Europe , Female , Ghana , Heart Disease Risk Factors , Humans , Logistic Models , Male , Middle Aged , Obesity/etiology , Predictive Value of Tests , ROC Curve , Reference Values , Sex Factors , Waist Circumference , Waist-Hip Ratio
2.
Nutr Diabetes ; 5: e181, 2015 Oct 19.
Article in English | MEDLINE | ID: mdl-26479315

ABSTRACT

BACKGROUND: ACAT-related enzyme 2 required for viability 1 (ARV1) is a putative lipid transporter of the endoplasmic reticulum that is conserved across eukaryotic species. The ARV1 protein contains a conserved N-terminal cytosolic zinc ribbon motif known as the ARV1 homology domain, followed by multiple transmembrane regions anchoring it in the ER. Deletion of ARV1 in yeast results in defective sterol trafficking, aberrant lipid synthesis, ER stress, membrane disorganization and hypersensitivity to fatty acids (FAs). We sought to investigate the role of Arv1 in mammalian lipid metabolism. METHODS: Homologous recombination was used to disrupt the Arv1 gene in mice. Animals were examined for alterations in lipid and lipoprotein levels, body weight, body composition, glucose tolerance and energy expenditure. RESULTS: Global loss of Arv1 significantly decreased total cholesterol and high-density lipoprotein cholesterol levels in the plasma. Arv1 knockout mice exhibited a dramatic lean phenotype, with major reductions in white adipose tissue (WAT) mass and body weight on a chow diet. This loss of WAT is accompanied by improved glucose tolerance, higher adiponectin levels, increased energy expenditure and greater rates of whole-body FA oxidation. CONCLUSIONS: This work identifies Arv1 as an important player in mammalian lipid metabolism and whole-body energy homeostasis.

3.
Gene Ther ; 21(7): 653-61, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24830434

ABSTRACT

Adipose tissue has a critical role in energy and metabolic homeostasis, but it is challenging to adapt techniques to modulate adipose function in vivo. Here we develop an in vivo, systemic method of gene transfer specifically targeting adipose tissue using adeno-associated virus (AAV) vectors. We constructed AAV vectors containing cytomegalovirus promoter-regulated reporter genes, intravenously injected adult mice with vectors using multiple AAV serotypes, and determined that AAV2/8 best targeted adipose tissue. Altering vectors to contain adiponectin promoter/enhancer elements and liver-specific microRNA-122 target sites restricted reporter gene expression to adipose tissue. As proof of efficacy, the leptin gene was incorporated into the adipose-targeted expression vector, package into AAV2/8 and administered intravenously to 9- to 10-week-old ob/ob mice. Phenotypic changes were measured over an 8-week period. Leptin mRNA and protein were expressed in adipose and leptin protein was secreted into plasma. Mice responded with reversal of weight gain, decreased hyperinsulinemia and improved glucose tolerance. AAV2/8-mediated systemic delivery of an adipose-targeted expression vector can replace a gene lacking in adipose tissue and correct a mouse model of human disease, demonstrating experimental application and therapeutic potential in disorders of adipose.


Subject(s)
Adipose Tissue/metabolism , Dependovirus/classification , Dependovirus/genetics , Gene Targeting/methods , Genetic Therapy/methods , Genetic Vectors/administration & dosage , 3' Untranslated Regions , Adiponectin/genetics , Adipose Tissue/virology , Animals , Disease Models, Animal , Gene Transfer Techniques , Genetic Vectors/genetics , Humans , Leptin/blood , Leptin/genetics , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Obesity/blood , Obesity/therapy , Organ Specificity
4.
Am J Physiol Endocrinol Metab ; 299(2): E249-57, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20484013

ABSTRACT

The excess accumulation of lipids in islets is thought to contribute to the development of diabetes in obesity by impairing beta-cell function. However, lipids also serve a nutrient function in islets, and fatty acids acutely increase insulin secretion. A better understanding of lipid metabolism in islets will shed light on complex effects of lipids on beta-cells. Adipose differentiation-related protein (ADFP) is localized on the surface of lipid droplets in a wide range of cells and plays an important role in intracellular lipid metabolism. We found that ADFP was highly expressed in murine beta-cells. Moreover, islet ADFP was increased in mice on a high-fat diet (3.5-fold of control) and after fasting (2.5-fold of control), revealing dynamic changes in ADFP in response to metabolic cues. ADFP expression was also increased by addition of fatty acids in human islets. The downregulation of ADFP in MIN6 cells by antisense oligonucleotide (ASO) suppressed the accumulation of triglycerides upon fatty acid loading (56% of control) along with a reduction in the mRNA levels of lipogenic genes such as diacylglycerol O-acyltransferase-2 and fatty acid synthase. Fatty acid uptake, oxidation, and lipolysis were also reduced by downregulation of ADFP. Moreover, the reduction of ADFP impaired the ability of palmitate to increase insulin secretion. These findings demonstrate that ADFP is important in regulation of lipid metabolism and insulin secretion in beta-cells.


Subject(s)
Insulin/biosynthesis , Islets of Langerhans/physiology , Lipid Metabolism/physiology , Membrane Proteins/physiology , Animals , Blood Glucose/metabolism , Blotting, Western , Cells, Cultured , Down-Regulation/physiology , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Lipolysis/physiology , Male , Mice , Mice, Inbred C57BL , Nutritional Physiological Phenomena , Perilipin-2 , RNA/biosynthesis , RNA/isolation & purification , Triglycerides/metabolism
5.
Int J Obes (Lond) ; 33(11): 1265-73, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19668253

ABSTRACT

BACKGROUND: Myostatin, also known as Growth and Differentiation Factor 8, is a secreted protein that inhibits muscle growth. Disruption of myostatin signaling increases muscle mass and decreases glucose, but it is unclear whether these changes are related. We treated mice on chow and high-fat diets with a soluble activin receptor type IIB (ActRIIB, RAP-031), which is a putative endogenous signaling receptor for myostatin and other ligands of the TGF-beta superfamily. RESULTS: After 4 weeks, RAP-031 increased lean and muscle mass, grip strength and contractile force. RAP-031 enhanced the ability of insulin to suppress glucose production under clamp conditions in high-fat fed mice, but did not significantly change insulin-mediated glucose disposal. The hepatic insulin-sensitizing effect of RAP-031 treatment was associated with increased adiponectin levels. RAP-031 treatment for 10 weeks further increased muscle mass and drastically reduced fat content in mice on either chow or high-fat diet. RAP-031 suppressed hepatic glucose production and increased peripheral glucose uptake in chow-fed mice. In contrast, RAP-031 suppressed glucose production with no apparent change in glucose disposal in high-fat-diet mice. CONCLUSION: Our findings show that disruption of ActRIIB signaling is a viable pharmacological approach for treating obesity and diabetes.


Subject(s)
Activin Receptors, Type II/metabolism , Insulin Resistance/physiology , Muscle, Skeletal/metabolism , Myostatin/metabolism , Obesity/metabolism , Animals , Case-Control Studies , Glucose Clamp Technique , Male , Mice , Mice, Inbred C57BL , Obesity/drug therapy , Obesity/physiopathology , Signal Transduction , Solubility
6.
Endocrinology ; 146(10): 4377-85, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16002520

ABSTRACT

Prader-Willi syndrome (PWS) has a biphasic clinical phenotype with failure to thrive in the neonatal period followed by hyperphagia and severe obesity commencing in childhood among other endocrinological and neurobehavioral abnormalities. The syndrome results from loss of function of several clustered, paternally expressed genes in chromosome 15q11-q13. PWS is assumed to result from a hypothalamic defect, but the pathophysiological basis of the disorder is unknown. We hypothesize that a fetal developmental abnormality in PWS leads to the neonatal phenotype, whereas the adult phenotype results from a failure in compensatory mechanisms. To address this hypothesis and better characterize the neonatal failure to thrive phenotype during postnatal life, we studied a transgenic deletion PWS (TgPWS) mouse model that shares similarities with the first stage of the human syndrome. TgPWS mice have fetal and neonatal growth retardation associated with profoundly reduced insulin and glucagon levels. Consistent with growth retardation, TgPWS mice have deregulated liver expression of IGF system components, as revealed by quantitative gene expression studies. Lethality in TgPWS mice appears to result from severe hypoglycemia after postnatal d 2 after depletion of liver glycogen stores. Consistent with hypoglycemia, TgPWS mice appear to have increased fat oxidation. Ghrelin levels increase in TgPWS reciprocally with the falling glucose levels, suggesting that the rise in ghrelin reported in PWS patients may be secondary to a perceived energy deficiency. Together, the data reveal defects in endocrine pancreatic function as well as glucose and hepatic energy metabolism that may underlie the neonatal phenotype of PWS.


Subject(s)
Hormones/blood , Prader-Willi Syndrome/physiopathology , Aging/physiology , Animals , Animals, Newborn , Body Weight , DNA Primers , Disease Models, Animal , Exons , Failure to Thrive , Fetus , Gene Deletion , Glucagon/blood , Insulin/blood , Liver Glycogen/metabolism , Mice , Mice, Transgenic , Prader-Willi Syndrome/metabolism , Reverse Transcriptase Polymerase Chain Reaction
7.
Horm Metab Res ; 34(11-12): 691-7, 2002.
Article in English | MEDLINE | ID: mdl-12660884

ABSTRACT

Consumption of a high-fat diet decreases hypothalamic neuropeptide Y (NPY) and increases proopiomelanocortin (POMC) and brown adipose uncoupling protein (UCP)-1 mRNA in obesity-resistant SWR/J but not obesity-prone C57Bl/6J mice. Although leptin was elevated in both strains in response to a high-fat diet, its role in the development of diet-induced obesity has remained unclear since insulin and other factors that affect similar tissue targets are altered. Thus, we administered recombinant leptin by subcutaneous infusion to chow-fed mice to mimic the changes in plasma leptin across its broad physiologic range. We observed strain differences in responsiveness to reduced and elevated leptin levels. A reduction in leptin during fasting evoked a greater response in C57Bl/6J mice by decreasing energy expenditure and thyroxin, increasing corticosterone and stimulating food intake and weight gain during refeeding. However, C57Bl/6J mice were less responsive to an increase in leptin in the fed state. Conversely, the leptin-mediated response to fasting was blunted in SWR/J mice, whereas an increase in leptin profoundly reduced food intake and body weight in SWR/J mice fed ad libitum. Sensitivity to fasting in C57Bl/6J mice was associated with higher hypothalamic NPY mRNA and reduced POMC and UCP-1 mRNA expression, while the robust response to high leptin levels in SWR/J mice was associated with suppression of NPY mRNA. These results indicate that differences in leptin responsiveness between strains might occur centrally or peripherally, leading to alteration in the patterns of food intake, thermogenesis and energy storage.


Subject(s)
Dietary Fats/metabolism , Fasting/blood , Leptin/blood , Obesity/metabolism , Animals , Body Weight/physiology , Dose-Response Relationship, Drug , Energy Metabolism/physiology , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Obesity/genetics , Recombinant Proteins , Signal Transduction , Species Specificity
8.
J Clin Invest ; 107(8): 1017-23, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11306605

ABSTRACT

Thyroid hormone thyroxine (T(4)) and tri-iodothyronine (T(3)) production is regulated by feedback inhibition of thyrotropin (TSH) and thyrotropin-releasing hormone (TRH) synthesis in the pituitary and hypothalamus when T(3) binds to thyroid hormone receptors (TRs) interacting with the promoters of the genes for the TSH subunit and TRH. All of the TR isoforms likely participate in the negative regulation of TSH production in vivo, but the identity of the specific TR isoforms that negatively regulate TRH production are less clear. To clarify the role of the TR-beta2 isoform in the regulation of TRH gene expression in the hypothalamic paraventricular nucleus, we examined preprothyrotropin-releasing hormone (prepro-TRH) expression in mice lacking the TR-beta2 isoform under basal conditions, after the induction of hypothyroidism with propylthiouracil, and in response to T(3) administration. Prepro-TRH expression was increased in hypothyroid wild-type mice and markedly suppressed after T(3) administration. In contrast, basal TRH expression was increased in TR-beta2-null mice to levels seen in hypothyroid wild-type mice and did not change significantly in response to induction of hypothyroidism or T(3) treatment. However, the suppression of TRH mRNA expression in response to leptin reduction during fasting was preserved in TR-beta2-null mice. Thus TR-beta2 is the key TR isoform responsible for T(3)-mediated negative-feedback regulation by hypophysiotropic TRH neurons.


Subject(s)
Paraventricular Hypothalamic Nucleus/metabolism , Receptors, Thyroid Hormone/physiology , Thyrotropin-Releasing Hormone/metabolism , Animals , Fasting , Leptin/metabolism , Mice , Mice, Knockout , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/cytology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/physiology , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Thyrotropin/metabolism , Thyrotropin-Releasing Hormone/genetics , Thyroxine/metabolism
9.
Trends Mol Med ; 7(5): 205-13, 2001 May.
Article in English | MEDLINE | ID: mdl-11325632

ABSTRACT

Obesity is highly prevalent in industralized countries and is increasing worldwide. It is also a major risk factor for type 2 diabetes, hypertension, coronary artery disease and certain cancers. An understanding of the regulation of eating behavior is pertinent to obesity, as the latter results from an imbalance between food consumption and energy expenditure. Leptin and other hormones regulate feeding and energy balance by modulating the expression of neuropeptides in the brain. Major efforts are underway to determine whether the peripheral and central pathways involved in the regulation of feeding behavior and energy balance could be targeted for the treatment of obesity.


Subject(s)
Eating/genetics , Feeding Behavior/physiology , Obesity/genetics , Animals , Body Weight , Cytokines/metabolism , Digestive System Physiological Phenomena , Humans , Leptin/metabolism , Models, Biological , Neurotransmitter Agents/metabolism , Obesity/therapy , Peptides/metabolism , Perception , Rats
10.
J Comp Neurol ; 432(1): 1-19, 2001 Mar 26.
Article in English | MEDLINE | ID: mdl-11241374

ABSTRACT

Cocaine- and amphetamine-regulated transcript (CART) is a recently described neuropeptide widely expressed in the rat brain. CART mRNA and peptides are found in hypothalamic sites such as the paraventricular nucleus (PVH), the supraoptic nucleus (SON), the lateral hypothalamic area (LHA), the dorsomedial nucleus of the hypothalamus (DMH), the arcuate nucleus (Arc), the periventricular nucleus (Pe), and the ventral premammillary nucleus (PMV). Intracerebroventricular administration of recombinant CART peptide decreases food intake and CART mRNA levels in the Arc are regulated by leptin. Leptin administration induces Fos expression in hypothalamic CART neurons in the PVH, the DMH, the Arc, and the PMV. In the current study, we used double label in situ hybridization histochemistry to investigate the potential direct action of leptin on hypothalamic CART neurons and to define the chemical identity of the hypothalamic CART neurons in the rat brain. We found that CART neurons in the Arc, DMH, and PMV express long form leptin-receptor mRNA, and the suppressor of cytokine signaling-3 (SOCS-3) mRNA after an acute dose of intravenous leptin. We also found that CART neurons in the parvicellular PVH, in the DMH and in the posterior Pe coexpress thyrotropin-releasing hormone (TRH) mRNA. CART neurons in the magnocellular PVH and in the SON coexpress dynorphin (DYN), and CART cell bodies in the LHA and in the posterior Pe coexpress melanin-concentrating hormone (MCH) and glutamic acid decarboxylase (GAD-67) mRNA. In the Arc, a few CART neurons coexpress neurotensin (NT) mRNA. In addition, we examined the distribution of CART immunoreactivity in the human hypothalamus. We found CART cell bodies in the PVH, in the SON, in the LHA, in the Arc (infundibular nucleus) and in the DMH. We also observed CART fibers throughout the hypothalamus, in the bed nucleus of the stria terminalis, and in the amygdala. Our results indicate that leptin directly acts on CART neurons in distinct nuclei of the rat hypothalamus. Furthermore, hypothalamic CART neurons coexpress neuropeptides involved in energy homeostasis, including MCH, TRH, DYN, and NT. The distribution of CART cell bodies and fibers in the human hypothalamus indicates that CART may also play a role in the regulation of energy homeostasis in humans.


Subject(s)
Gene Expression Regulation , Hypothalamus/metabolism , Leptin/pharmacology , Nerve Tissue Proteins/genetics , Neurons/metabolism , Adult , Aged , Animals , Feeding Behavior/drug effects , Female , Gene Expression Regulation/drug effects , Humans , Hypothalamic Hormones/genetics , Hypothalamus/cytology , Immunohistochemistry , Injections, Intraventricular , Intracellular Signaling Peptides and Proteins , Male , Melanins/genetics , Middle Aged , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/pharmacology , Neurons/cytology , Neuropeptides/analysis , Neuropeptides/genetics , Neurotransmitter Agents/analysis , Neurotransmitter Agents/genetics , Orexins , Organ Specificity , Pituitary Hormones/genetics , Protein Biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Species Specificity , Transcription, Genetic
11.
Nature ; 409(6818): 307-12, 2001 Jan 18.
Article in English | MEDLINE | ID: mdl-11201732

ABSTRACT

Diabetes mellitus is a chronic disease that leads to complications including heart disease, stroke, kidney failure, blindness and nerve damage. Type 2 diabetes, characterized by target-tissue resistance to insulin, is epidemic in industrialized societies and is strongly associated with obesity; however, the mechanism by which increased adiposity causes insulin resistance is unclear. Here we show that adipocytes secrete a unique signalling molecule, which we have named resistin (for resistance to insulin). Circulating resistin levels are decreased by the anti-diabetic drug rosiglitazone, and increased in diet-induced and genetic forms of obesity. Administration of anti-resistin antibody improves blood sugar and insulin action in mice with diet-induced obesity. Moreover, treatment of normal mice with recombinant resistin impairs glucose tolerance and insulin action. Insulin-stimulated glucose uptake by adipocytes is enhanced by neutralization of resistin and is reduced by resistin treatment. Resistin is thus a hormone that potentially links obesity to diabetes.


Subject(s)
Diabetes Mellitus, Type 2/complications , Diabetes Mellitus/etiology , Hormones, Ectopic/physiology , Hormones/physiology , Intercellular Signaling Peptides and Proteins , Obesity , Proteins , Thiazolidinediones , 3T3 Cells , Adipocytes/metabolism , Amino Acid Sequence , Animals , Base Sequence , Chromosomes, Human, Pair 19 , Cloning, Molecular , DNA , Diabetes Mellitus/blood , Diabetes Mellitus/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Diet , Female , Gene Expression Regulation/drug effects , Glucose Intolerance , Hormones/genetics , Hormones/metabolism , Hormones, Ectopic/genetics , Hormones, Ectopic/metabolism , Humans , Hypoglycemic Agents/pharmacology , Insulin Antagonists , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Nerve Growth Factor , Neutralization Tests , Resistin , Rosiglitazone , Signal Transduction , Thiazoles/pharmacology
12.
Regul Pept ; 92(1-3): 1-7, 2000 Aug 25.
Article in English | MEDLINE | ID: mdl-11024558

ABSTRACT

Leptin is produced mainly by adipose tissue and has been shown to regulate feeding, energy balance and neuroendocrine function. Regulation of energy homeostasis by leptin is thought to be mediated by hypothalamic neuropeptides, at least in adult rodents. The neonatal period is a critical stage of development during which mammals have to optimize caloric intake to support growth and development, as well as maintain body temperature. It is likely that leptin is involved in the transition from preweaning to adult metabolism. To test this hypothesis, we compared the effect of leptin treatment on body weight and adiposity between neonatal and adult mice. We also determined whether well known hypothalamic neuropeptide targets, e.g. neuropeptide Y (NPY), proopiomelanocortin (POMC), agouti-related peptide (AGRP) and cocaine and amphetamine-regulated transcript (CART) were regulated in a pattern consistent with their presumed roles as mediators of leptin action. Once daily intraperitoneal leptin injection for 7 days did not alter body weight, fat content or expression of hypothalamic neuropeptide mRNAs in 10-day-old mice. In contrast, leptin decreased body weight and adiposity, increased CART and suppressed NPY and AGRP mRNA expression in adult mice. These results are consistent with previous studies showing that the timing of leptin's anorectic action develops after weaning. Furthermore, the association between leptin's ability to influence body weight in adult mice but not in neonates, and the regulation of hypothalamic neuropeptide mRNA expression, is consistent with the view that these peptides mediate leptin's effects on energy balance.


Subject(s)
Leptin/metabolism , Nerve Tissue Proteins/genetics , Neuropeptide Y/genetics , Pro-Opiomelanocortin/genetics , Proteins/genetics , Agouti-Related Protein , Animals , Body Weight/drug effects , Corticosterone/metabolism , Energy Metabolism/drug effects , Fats/metabolism , Gene Expression Regulation/drug effects , Hypothalamus/metabolism , Insulin/metabolism , Intercellular Signaling Peptides and Proteins , Leptin/administration & dosage , Male , Mice , Mice, Inbred C57BL , RNA, Messenger , Triglycerides/metabolism
13.
Trends Endocrinol Metab ; 11(8): 327-32, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10996528

ABSTRACT

The discovery of leptin in the mid-1990s has focused attention on the role of proteins secreted by adipose tissue. Leptin has profound effects on appetite and energy balance, and is also involved in the regulation of neuroendocrine and immune function. Sex steroid and glucocorticoid metabolism in adipose tissue has been implicated as a determinant of body fat distribution and cardiovascular risk. Other adipose products, for example, proinflammatory cytokines, complement factors and components of the coagulation/fibrinolytic cascade, may mediate the metabolic and cardiovascular complications associated with obesity.


Subject(s)
Adipose Tissue/physiology , Endocrine Glands/physiology , Hormones/physiology , Adipose Tissue/metabolism , Animals , Hormones/biosynthesis , Humans
14.
Front Neuroendocrinol ; 21(3): 263-307, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10882542

ABSTRACT

The discovery of leptin has enhanced understanding of the interrelationship between adipose energy stores and neuronal circuits in the brain involved in energy balance and regulation of the neuroendocrine axis. Leptin levels are dependent on the status of fat stores as well as changes in energy balance as a result of fasting and overfeeding. Although leptin was initially thought to serve mainly as an anti-satiety hormone, recent studies have shown that it mediates the adaptation to fasting. Furthermore, leptin has been implicated in the regulation of the reproductive, thyroid, growth hormone, and adrenal axes, independent of its role in energy balance. Although it is widely known that leptin acts on hypothalamic neuronal targets to regulate energy balance and neuroendocrine function, the specific neuronal populations mediating leptin action on feeding behavior and autonomic and neuroendocrine function are not well understood. In this review, we have discussed how leptin engages arcuate hypothalamic neurons expressing putative orexigenic peptides, e.g., neuropeptide Y and agouti-regulated peptide, and anorexigenic peptides, e.g., pro-opiomelanocortin (precursor of alpha-melanocyte-stimulating hormone) and cocaine- and amphetamine-regulated transcript. We show that leptin's effects on energy balance and the neuroendocrine axis are mediated by projections to other hypothalamic nuclei, e.g., paraventricular, lateral, and perifornical areas, as well as other sites in the brainstem, spinal cord, and cortical and subcortical regions.


Subject(s)
Leptin/physiology , Neurosecretory Systems/physiology , Animals , Biological Transport , Brain/metabolism , Energy Metabolism/physiology , Homeostasis/physiology , Humans , Leptin/metabolism
15.
J Comp Neurol ; 423(2): 261-81, 2000 Jul 24.
Article in English | MEDLINE | ID: mdl-10867658

ABSTRACT

Leptin has profound effects on food intake, body weight, and neuroendocrine status. The lack of leptin results in hormonal and metabolic alterations and a dramatic increase in body weight. Leptin acts in the brain, especially in the hypothalamus; however, the central nervous system sites that respond to leptin have not been examined comprehensively. In this study, we explored systematically the distribution of leptin-activated neurons throughout the rat brain. Furthermore, we investigated the chemical identity of subsets of these leptin-activated cells. Fos-like immunoreactivity (Fos-IR) was investigated in the rat brain after two different doses of leptin (1.0 mg/kg and 5.0 mg/kg) at 2 hours and 6 hours after injections. The induction of Fos-IR was observed in hypothalamic nuclei, including the paraventricular nucleus (PVH), the retrochiasmatic area (RCA), the ventromedial nucleus (VMH), the dorsomedial nucleus (DMH), the arcuate nucleus (Arc), and the ventral premammillary nucleus (PMV). In addition, leptin-induced Fos-IR was found in several nuclei of the brainstem, including the superior lateral and external lateral subdivisions of the parabrachial nucleus (slPB and elPB, respectively), the supragenual nucleus, and the nucleus of the solitary tract (NTS). By using double-labeling immunohistochemistry or immunohistochemistry coupled with in situ hybridization, leptin-activated neurons were found that contained cocaine- and amphetamine-regulated transcript mRNA in several hypothalamic nuclei, including the RCA, Arc, DMH, and PMV. In the Arc and DMH, leptin-induced Fos-IR was observed in neurons that expressed neurotensin mRNA. Dynorphin neurons in the VMH and in the Arc also expressed Fos-IR. In the brainstem, we found that cholecystokinin neurons in the slPB and glucagon-like peptide-1 neurons in the NTS were activated by leptin. We also investigated the coexpression of Fos-IR and the long form of the leptin receptor (OBRb) mRNA. We found double-labeled neurons surrounding the median eminence and in the RCA, Arc, VMH, DMH, and PMV. However, in brainstem sites, very little OBRb mRNA was found; thus, there were very few double-labeled cells. These results suggest that leptin stimulates brain pathways containing neuropeptides that are involved in the regulation of energy balance, autonomic homeostasis, and neuroendocrine status.


Subject(s)
Brain Chemistry/physiology , Brain/metabolism , Caenorhabditis elegans Proteins , Eating/physiology , Leptin/metabolism , Neurons/chemistry , Rats, Sprague-Dawley/metabolism , Receptors, Cell Surface , Animals , Brain/cytology , Carrier Proteins/genetics , Cholecystokinin/genetics , Energy Metabolism/physiology , Enkephalins/genetics , Homeostasis/physiology , Immunohistochemistry , In Situ Hybridization , Leptin/pharmacology , Male , Membrane Glycoproteins/genetics , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/metabolism , Neurotensin/genetics , Protein Precursors/genetics , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley/anatomy & histology , Receptors, Leptin , Receptors, Notch , Time Factors
16.
Annu Rev Physiol ; 62: 413-37, 2000.
Article in English | MEDLINE | ID: mdl-10845097

ABSTRACT

The discovery of the adipose-derived hormone leptin has generated enormous interest in the interaction between peripheral signals and brain targets involved in the regulation of feeding and energy balance. Plasma leptin levels correlate with fat stores and respond to changes in energy balance. It was initially proposed that leptin serves a primary role as an anti-obesity hormone, but this role is commonly thwarted by leptin resistance. Leptin also serves as a mediator of the adaptation to fasting, and this role may be the primary function for which the molecule evolved. There is increasing evidence that leptin has systemic effects apart from those related to energy homeostasis, including regulation of neuroendocrine and immune function and a role in development.


Subject(s)
Leptin/physiology , Adipose Tissue/physiology , Animals , Humans , Leptin/genetics
18.
Anat Embryol (Berl) ; 200(5): 509-19, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10526019

ABSTRACT

The spatial and temporal distribution of apoptosis in the dorsal midline of the developing chick hindbrain was examined in relation to the development of the neuroepithelium and neural crest using scanning and transmission electron microscopy, immunocytochemistry and in situ hybridization. The pattern of TUNEL labeling and Slug expression in the dorsal midline at stages 10 and 11 differed from that at stages 12-15. At stages 10 and 11, TUNEL labeling and Slug expression were observed in the dorsal part of location II of rhombomere 1/2 (i.e., between the surface ectoderm and the neuroepithelium), but from stage 12 onward, they were observed in both the dorsal and ventral parts of location II. The implication is that whereas apoptosis may be restricted to a subpopulation of the early migrating neural crest at stages 10 and 11, it presumably occurs in subpopulations of both neural crest and neuroepithelial cells from stage 12 onward. Furthermore, as judged by the pattern of TUNEL labeling and Slug expression in r3 and r5, apoptosis in these two rhombomeres likely occurs in subpopulations of both neural crest and neuroepithelial cells. The eminence present in location I of r1/r2 between stages 10 and 12 consisted of both neural crest and neuroepithelial cells. These cells gradually underwent apoptosis until stage 12, when the eminence disappeared in most embryos. The formation of the inner (neuroepithelial) aspect of the hindbrain roof plate involved both cell migration from adjacent neuroepithelium and an alteration in the shapes of the cells, such that cells with flattened surfaces eventually lined the roof plate. During these processes, some of the neuroepithelial cells underwent apoptosis (i.e., in location IV). The results of this study thus demonstrate that subpopulations of both neuroepithelial and neural crest cells may be involved in programmed cell death in the hindbrain. Additionally, apoptosis in the hindbrain contributes significantly to morphogenetic thinning during roof plate formation.


Subject(s)
Apoptosis/physiology , Neural Crest/chemistry , Neural Crest/embryology , Rhombencephalon/chemistry , Rhombencephalon/embryology , Animals , Chick Embryo , In Situ Nick-End Labeling , Microscopy, Electron , Microscopy, Electron, Scanning , Rhombencephalon/ultrastructure , Snail Family Transcription Factors , Sulfotransferases/analysis , Transcription Factors/analysis
19.
Endocrinology ; 140(11): 4923-31, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10537115

ABSTRACT

Leptin acts on specific populations of hypothalamic neurons to regulate feeding behavior, energy expenditure, and neuroendocrine function. It is not known, however, whether the same neural circuits mediate leptin action across its full biologic dose-response curve, which extends over a broad range, from low levels seen during starvation to high levels characteristic of obesity. Here, we show that the characteristic fall in leptin with fasting causes a rise in neuropeptide Y (NPY) messenger RNA (mRNA), as well as a fall in POMC and cocaine and amphetamine-regulated transcript (CART) mRNAs. Sc infusion of leptin sufficient to maintain plasma levels within the physiologic range during the fast prevents changes in the expression of these peptides, as well as changes in neuroendocrine function, demonstrating that multiple neural circuits are highly sensitive to small changes in leptin within its low physiologic range. In contrast, a modest elevation of plasma leptin above the normal fed range by constant sc infusion, which produced marked reduction in food intake and body weight, decreased NPY mRNA in the arcuate hypothalamic nucleus but did not affect the levels of mRNAs encoding the anorexigenic peptides alpha-MSH, CART or CRH. These results suggest that the dose response characteristics of leptin on hypothalamic target neurons at the level of mRNA expression are variable, with some neurons (e.g. NPY) responding across a broad dose range and others (e.g. POMC and CART) showing a limited response within the low range. These results further suggest that the central targets of leptin that mediate the transition from starvation to the fed state may be distinct from those that mediate the response to overfeeding and obesity.


Subject(s)
Hypothalamus/physiology , Leptin/physiology , Neurons/physiology , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Body Weight/drug effects , Corticotropin-Releasing Hormone/genetics , Eating/drug effects , Fasting , Gene Expression Regulation/drug effects , Leptin/administration & dosage , Leptin/deficiency , Male , Nerve Tissue Proteins/genetics , Neuropeptide Y/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , alpha-MSH/genetics
20.
Neuron ; 23(4): 775-86, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10482243

ABSTRACT

Recent studies have reinforced the view that the lateral hypothalamic area (LHA) regulates food intake and body weight. We identified leptin-sensitive neurons in the arcuate nucleus of the hypothalamus (Arc) that innervate the LHA using retrograde tracing with leptin administration. We found that retrogradely labeled cells in the Arc contained neuropeptide Y (NPY) mRNA or proopiomelanocortin (POMC) mRNA. Following leptin administration, NPY cells in the Arc did not express Fos but expressed suppressor of cytokine signaling-3 (SOCS-3) mRNA. In contrast, leptin induced both Fos and SOCS-3 expression in POMC neurons, many of which also innervated the LHA. These findings suggest that leptin directly and differentially engages NPY and POMC neurons that project to the LHA, linking circulating leptin and neurons that regulate feeding behavior and body weight homeostasis.


Subject(s)
Arcuate Nucleus of Hypothalamus/physiology , Hypothalamic Area, Lateral/physiology , Neurons/physiology , Neuropeptide Y/physiology , Pro-Opiomelanocortin/physiology , Proteins/physiology , Repressor Proteins , Transcription Factors , Animals , Arcuate Nucleus of Hypothalamus/cytology , Body Weight/physiology , Cell Count , Eating/physiology , Homeostasis/physiology , Hypothalamic Area, Lateral/cytology , Immunohistochemistry , In Situ Hybridization , Leptin , Male , Neural Pathways/cytology , Neural Pathways/physiology , Protein Biosynthesis , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-fos/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...